1
|
Shu X, Chen Z, Wang H, Xu B, Liu L, Zhang J, Zheng X, Chen J. Cloning, phylogenetic analysis, tissue expression profiling, and functional roles of NPC1L1 in chickens, quails, and ducks. Poult Sci 2025; 104:105032. [PMID: 40106905 PMCID: PMC11964625 DOI: 10.1016/j.psj.2025.105032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 03/01/2025] [Accepted: 03/13/2025] [Indexed: 03/22/2025] Open
Abstract
The Niemann-Pick C1-Like 1 (NPC1L1) protein, primarily expressed in the epithelial cells of the small intestine, is essential for cholesterol absorption from both dietary intake and biliary secretion. Despite this conserved function across mammals, the full-length coding sequence of NPC1L1 remains uncharacterized in key avian models including chicken (Gallus gallus), quail (Coturnix japonica), and duck (Anas platyrhynchos). In this study, we successfully cloned the full NPC1L1 mRNA sequence in chicken, quail, and duck, including the entire 5' and 3' untranslated regions, utilizing rapid amplification of cDNA ends methods. Phylogenetic analysis across 12 species, comprising four avian and eight representative mammalian species, revealed that the NPC1L1 sequences in the main poultry species exhibit a high degree of similarity. Despite the phylogenetic divergence of poultry NPC1L1 sequences from their mammalian counterparts, protein sequence alignment revealed that the cholesterol-sensing peptides of NPC1L1 are conserved across all species examined in this study. These findings imply that the NPC1L1 in poultry may also play a role in cholesterol transport. Analysis of tissue gene expression profiles in chickens, quails, and ducks indicated that NPC1L1 is predominantly expressed in the duodenum, jejunum, and liver. Additionally, experiments on medium-to-cell cholesterol transit in primary intestinal epithelial cells confirmed that chicken NPC1L1 is capable of efficiently transporting cholesterol into cells. Further experiments are required to elucidate the biological function of poultry NPC1L1. In summary, this study successfully cloned the full-length sequence of NPC1L1 from chickens, quails, and ducks, and conducted a comprehensive analysis of their evolutionary history and expression patterns. This research establishes a foundation for future investigations into the role of poultry NPC1L1 in cholesterol transport.
Collapse
Affiliation(s)
- Xin Shu
- Jiangsu Key Laboratory of Sericultural Biology and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, China
| | - Ziwei Chen
- Jiangsu Key Laboratory of Sericultural Biology and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, China
| | - Hui Wang
- Jiangsu Key Laboratory of Sericultural Biology and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, China
| | - Bingjie Xu
- Jiangsu Key Laboratory of Sericultural Biology and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, China
| | - Li Liu
- Jiangsu Key Laboratory of Sericultural Biology and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, China
| | - Jilong Zhang
- Jiangsu Key Laboratory of Sericultural Biology and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, China
| | - Xiaotong Zheng
- Jiangsu Key Laboratory of Sericultural Biology and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, China; Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang, China
| | - Jianfei Chen
- Jiangsu Key Laboratory of Sericultural Biology and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, China; Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang, China.
| |
Collapse
|
2
|
Chen X, Memory Kunda LS, Li X, Wang N, Huang Y, Hao Y, He Q, Liao W, Chen J. A Comprehensive Review of Beneficial Effects of Phytosterols on Glycolipid Metabolism and Related Mechanisms. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:3826-3841. [PMID: 39927454 DOI: 10.1021/acs.jafc.4c10375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2025]
Abstract
Phytosterols are widely distributed in various plant foods, such as nuts, grains, vegetables, and so on. Phytosterols have been broadly applied in functional foods, supplements, and pharmaceutical products due to their excellent cholesterol-lowering effect. Besides the cholesterol-lowering effect, recently, phytosterols have been found to exert a beneficial effect on glycolipid metabolism, which contributes to multiple metabolic diseases, such as diabetes, cardiovascular disease, and fatty liver. Constant development of new drugs with a single target fails to effectively curb the occurrence of metabolic diseases and complications, such as multiple organ damage, and phytosterols attract special attention due to varieties of biological activities, especially the regulation of glycolipid metabolism through multiple targets. Present review gives a comprehensive review of the effects of phytosterols on glycolipid metabolism and related mechanism. We also review the promising update of phytosterol in the treatment of two major metabolic diseases, including diabetes and nonalcohol fatty liver disease. This review can help to extend the understanding of the potential of phytosterols for mixed dyslipidemia and related metabolic diseases.
Collapse
Affiliation(s)
- Xiao Chen
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Lwara Sophie Memory Kunda
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Xinyang Li
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Nan Wang
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Yangjia Huang
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Yuting Hao
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Qi He
- School of Public Health, Southern Medical University, Guangzhou, Guangdong 510640, China
| | - Wenzhen Liao
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Jinyuan Chen
- Institute of Scientific Research, Southern Medical University, Guangzhou 510515, China
- TCM-Integrated Hospital, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
3
|
Oh S, Cho KH, Kim MC, Sim DS, Hong YJ, Kim JH, Ahn Y, Lee SY, Shin MH, Kim W, Jeong MH. Ten-year trends in lipid management among patients after myocardial infarction in South Korea. PLoS One 2024; 19:e0304710. [PMID: 39361921 PMCID: PMC11449489 DOI: 10.1371/journal.pone.0304710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 05/17/2024] [Indexed: 10/05/2024] Open
Abstract
BACKGROUND Dyslipidemia is an important risk factor for acute myocardial infarction. However, real-world data on its prevalence and lipid management trends for Korean patients with acute myocardial infarction are limited. This study aimed to determine the 10-year temporal trends in dyslipidemia prevalence and lipid management in this patient population. METHODS AND FINDINGS The study used a merged database of two nationwide observational cohorts (2011-2020) that included 26,751 participants. The primary endpoints were the achievement rates of the (1) absolute low-density lipoprotein cholesterol (LDL-C) target of <70 mg/dL (<1.8 mmol/L), (2) relative LDL-C target reduction of >50% from the baseline, (3) absolute or relative LDL-C target (American target), and (4) both absolute and relative LDL-C targets (European target). The dyslipidemia prevalence increased from 11.1% to 17.1%, whereas the statin prescription rate increased from 92.9% to 97.0% from 2011 to 2020. The rate of high-intensity statin use increased from 12.80% in 2012 to 69.30% in 2020. The rate of ezetimibe use increased from 4.50% in 2016 to 22.50% in 2020. The high-intensity statin and ezetimibe prescription rates (0.20% to 9.30% from 2016 to 2020) increased gradually. The absolute and relative LDL-C target achievement rates increased from 41.4% and 20.8% in 2012 to 62.5% and 39.5% in 2019, respectively. The American (45.7% in 2012 to 68.6% in 2019) and European (16.5% in 2012 to 33.8% in 2019) target achievement rates also increased. CONCLUSIONS The adoption of lipid management guidelines in clinical practice has improved. However, continued efforts are needed to reduce the risk of recurrent ischemic events.
Collapse
Affiliation(s)
- Seok Oh
- Department of Cardiology, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Kyung Hoon Cho
- Department of Cardiology, Chonnam National University Hospital, Gwangju, Republic of Korea
- Department of Cardiology, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Min Chul Kim
- Department of Cardiology, Chonnam National University Hospital, Gwangju, Republic of Korea
- Department of Cardiology, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Doo Sun Sim
- Department of Cardiology, Chonnam National University Hospital, Gwangju, Republic of Korea
- Department of Cardiology, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Young Joon Hong
- Department of Cardiology, Chonnam National University Hospital, Gwangju, Republic of Korea
- Department of Cardiology, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Ju Han Kim
- Department of Cardiology, Chonnam National University Hospital, Gwangju, Republic of Korea
- Department of Cardiology, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Youngkeun Ahn
- Department of Cardiology, Chonnam National University Hospital, Gwangju, Republic of Korea
- Department of Cardiology, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Sang Yeub Lee
- Chung-Ang University Gwangmyeong Hospital and Department of Internal Medicine, Division of Cardiology, Chung-Ang University College of Medicine, Gwangmyeong, Republic of Korea
| | - Min-Ho Shin
- Department of Preventive Medicine, Chonnam National University Medical School, Hwasun, Republic of Korea
| | - Weon Kim
- Department of Internal Medicine, Division of Cardiology, Kyung Hee University Hospital, Kyung Hee University, Seoul, Republic of Korea
| | - Myung Ho Jeong
- Department of Cardiology, Chonnam National University Hospital, Gwangju, Republic of Korea
- Department of Cardiology, Chonnam National University Medical School, Gwangju, Republic of Korea
- Department of Cardiology, Gwangju Veterans Hospital, Gwangju, Republic of Korea
| |
Collapse
|
4
|
Aguchem RN, Okagu IU, Okorigwe EM, Uzoechina JO, Nnemolisa SC, Ezeorba TPC. Role of CETP, PCSK-9, and CYP7-alpha in cholesterol metabolism: Potential targets for natural products in managing hypercholesterolemia. Life Sci 2024; 351:122823. [PMID: 38866219 DOI: 10.1016/j.lfs.2024.122823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 06/03/2024] [Accepted: 06/07/2024] [Indexed: 06/14/2024]
Abstract
Cardiovascular diseases (CVDs) are a leading cause of mortality worldwide, primarily affecting the heart and blood vessels, with atherosclerosis being a major contributing factor to their onset. Epidemiological and clinical studies have linked high levels of low-density lipoprotein (LDL) emanating from distorted cholesterol homeostasis as its major predisposing factor. Cholesterol homeostasis, which involves maintaining the balance in body cholesterol level, is mediated by several proteins or receptors, transcription factors, and even genes, regulating cholesterol influx (through dietary intake or de novo synthesis) and efflux (by their conversion to bile acids). Previous knowledge about CVDs management has evolved around modulating these receptors' activities through synthetic small molecules/antibodies, with limited interest in natural products. The central roles of the cholesteryl ester transfer protein (CETP), proprotein convertase subtilisin/kexin type 9 (PCSK9), and cytochrome P450 family 7 subfamily A member 1 (CYP7A1), among other proteins or receptors, have fostered growing scientific interests in understanding more on their regulatory activities and potential as drug targets. We present up-to-date knowledge on the contributions of CETP, PCSK9, and CYP7A1 toward CVDs, highlighting the clinical successes and failures of small molecules/antibodies to modulate their activities. In recommendation for a new direction to improve cardiovascular health, we have presented recent findings on natural products (including functional food, plant extracts, phytochemicals, bioactive peptides, and therapeutic carbohydrates) that also modulate the activities of CETP, PCSK-9, and CYP7A1, and emphasized the need for more research efforts redirected toward unraveling more on natural products potentials even at clinical trial level for CVD management.
Collapse
Affiliation(s)
- Rita Ngozi Aguchem
- Department of Biochemistry, Faculty of Biological Sciences, University of Nigeria, Enugu State 410001, Nigeria
| | - Innocent Uzochukwu Okagu
- Department of Biochemistry, Faculty of Biological Sciences, University of Nigeria, Enugu State 410001, Nigeria
| | - Ekezie Matthew Okorigwe
- Department of Biochemistry, Faculty of Biological Sciences, University of Nigeria, Enugu State 410001, Nigeria; Department of Chemistry and Biochemistry, College of Sciences, University of Notre Dame, 46556 Notre Dame, IN, United States
| | - Jude Obiorah Uzoechina
- Department of Biochemistry, Faculty of Biological Sciences, University of Nigeria, Enugu State 410001, Nigeria; Department of Biochemistry and Molecular Biology, Institute of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Shenzhen University Town, Shenzhen, PR China
| | | | - Timothy Prince Chidike Ezeorba
- Department of Biochemistry, Faculty of Biological Sciences, University of Nigeria, Enugu State 410001, Nigeria; Department of Genetics and Biotechnology, Faculty of Biological Sciences, University of Nigeria, Enugu State 410001, Nigeria; Department of Environmental Health and Risk Management, College of Life and Environmental Sciences, University of Birmingham, Edgbaston B15 2TT, United Kingdom.
| |
Collapse
|
5
|
Xu C, Fu F, She Y, Xu C. NPC1L1 Plays a Novel Role in Nonalcoholic Fatty Liver Disease. ACS OMEGA 2023; 8:48586-48589. [PMID: 38162748 PMCID: PMC10753569 DOI: 10.1021/acsomega.3c07337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 11/24/2023] [Accepted: 11/28/2023] [Indexed: 01/03/2024]
Abstract
Niemann-Pick C1-Like 1 (NPC1L1) is a key protein in the transport of cholesterol, which exists in the brush marginal membrane of the intestinal epithelial cells and the timid duct membrane of the liver. It affects cholesterol absorption and plasma low-density lipoprotein levels. Cholesterol is both an important component of the cell membrane and a precursor of bile acid and steroid hormone synthesis. Abnormal cholesterol metabolism is closely related to nonalcoholic steatohepatitis (NASH). NASH can progress to fibrosis and cirrhosis, with serious consequences. NPC1L1 is involved in the regulation of cholesterol and lipid metabolism and plays an important role in maintaining the balance of cholesterol metabolism in the body. It also plays an important role in some metabolic diseases such as nonalcoholic fatty liver disease, obesity, and hypercholesterolemia. Therefore, it is necessary to elucidate the molecular pathological mechanism of NPC1L1 in the regulation of cholesterol metabolism and the occurrence and development of NASH, which can provide a target for the development of novel drugs for the treatment of NASH and other diseases. More importantly, it helps to accelerate the development of drugs that regulate lipid metabolism at multiple levels and reduce liver steatosis, which is extremely important for the prevention and treatment of NASH and related severe metabolic diseases.
Collapse
Affiliation(s)
- ChongLi Xu
- College
of Medical Technology, Chongqing Medical
and Pharmaceutical College, 82 Daxuecheng Road, Chongqing 401331, PR China
| | - Fengyang Fu
- College
of Medical Technology, Chongqing Medical
and Pharmaceutical College, 82 Daxuecheng Road, Chongqing 401331, PR China
| | - Yuhan She
- College
of Medical Technology, Chongqing Medical
and Pharmaceutical College, 82 Daxuecheng Road, Chongqing 401331, PR China
| | - ChongBo Xu
- School
of Biology and Agriculture, Shaoguan University, Shaoguan 512005, PR China
| |
Collapse
|
6
|
Oh EY, Haam CE, Choi S, Byeon S, Choi SK, Lee YH. Ezetimibe Induces Vasodilation in Rat Mesenteric Resistance Arteries through Inhibition of Extracellular Ca 2+ Influx. Int J Mol Sci 2023; 24:13992. [PMID: 37762296 PMCID: PMC10531054 DOI: 10.3390/ijms241813992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 09/07/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
Ezetimibe is a lipid-lowering agent that selectively inhibits cholesterol absorption by binding to the Niemann-Pick C1-like 1 (NPC1L1) protein. Although it is well known that administration of ezetimibe in hypercholesterolemia patients reduces the risk of cardiovascular events through attenuation of atherosclerosis, studies on the direct effect of ezetimibe on vascular function are not sufficient. The aim of the present study was to investigate the vascular effects of ezetimibe in rat mesenteric arteries. In the present study, 12-week-old male Sprague Dawley rats were used. After the rats were sacrificed, the second branches of the mesenteric arteries were isolated and cut into 2-3 mm segments and mounted in a multi-wire myography system to measure isometric tension. Ezetimibe reduced vasoconstriction induced by U46619 (500 nM) in endothelium-intact and endothelium-denuded arteries. Ezetimibe-induced vasodilation was not affected by the endothelial nitric oxide synthase (eNOS) inhibitor Nω-Nitro-L-arginine (L-NNA, 300 μM) or the non-selective potassium channel blocker, tetraethylammonium (TEA, 10 mM). Moreover, ezetimibe also completely blocked the contraction induced by an increase in external calcium concentration. Ezetimibe significantly reduced vascular contraction induced by L-type Ca2+ channel activator (Bay K 8644, 30 nM). Treatment with ezetimibe decreased the phosphorylation level of 20 kDa myosin light chain (MLC20) in vascular smooth muscle cells. In the present study, we found that ezetimibe has a significant vasodilatory effect in rat mesenteric resistance arteries. These results suggest that ezetimibe may have beneficial cardiovascular effects beyond its cholesterol-lowering properties.
Collapse
Affiliation(s)
| | | | | | | | - Soo-Kyoung Choi
- Department of Physiology, Yonsei University College of Medicine, 50 Yonseiro, Seodaemun-gu, Seoul 03722, Republic of Korea; (E.Y.O.); (C.E.H.); (S.C.); (S.B.)
| | - Young-Ho Lee
- Department of Physiology, Yonsei University College of Medicine, 50 Yonseiro, Seodaemun-gu, Seoul 03722, Republic of Korea; (E.Y.O.); (C.E.H.); (S.C.); (S.B.)
| |
Collapse
|
7
|
Colletti A, Fratter A, Pellizzato M, Cravotto G. Nutraceutical Approaches to Dyslipidaemia: The Main Formulative Issues Preventing Efficacy. Nutrients 2022; 14:nu14224769. [PMID: 36432457 PMCID: PMC9696395 DOI: 10.3390/nu14224769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 10/29/2022] [Accepted: 11/04/2022] [Indexed: 11/16/2022] Open
Abstract
Currently, the nutraceutical approach to treat dyslipidaemia is increasing in use, and in many cases is used by physicians as the first choice in the treatment of patients with borderline values. Nutraceuticals represent an excellent opportunity to treat the preliminary conditions not yet showing the pathological signs of dyslipidaemia. Their general safety, the patient's confidence, the convincing proof of efficacy and the reasonable costs prompted the market of new preparations. Despite this premise, many nutraceutical products are poorly formulated and do not meet the minimum requirements to ensure efficacy in normalizing blood lipid profiles, promoting cardiovascular protection, and normalizing disorders of glycemic metabolism. In this context, bioaccessibility and bioavailability of the active compounds is a crucial issue. Little attention is paid to the proper formulations needed to improve the overall bioavailability of the active molecules. According to these data, many products prove to be insufficient to ensure full enteric absorption. The present review analysed the literature in the field of nutraceuticals for the treatment of dyslipidemia, focusing on resveratrol, red yeast rice, berberine, and plant sterols, which are among the nutraceuticals with the greatest formulation problems, highlighting bioavailability and the most suitable formulations.
Collapse
Affiliation(s)
- Alessandro Colletti
- Department of Science and Drug Technology, University of Turin, 10124 Turin, Italy
- Italian Society of Nutraceutical Formulators (SIFNut), 31033 Treviso, Italy
| | - Andrea Fratter
- Italian Society of Nutraceutical Formulators (SIFNut), 31033 Treviso, Italy
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, 35122 Padua, Italy
| | - Marzia Pellizzato
- Italian Society of Nutraceutical Formulators (SIFNut), 31033 Treviso, Italy
| | - Giancarlo Cravotto
- Department of Science and Drug Technology, University of Turin, 10124 Turin, Italy
- Italian Society of Nutraceutical Formulators (SIFNut), 31033 Treviso, Italy
- Correspondence: ; Tel.: +39-011-670-7103
| |
Collapse
|
8
|
Recent advances in the screening methods of NPC1L1 inhibitors. Biomed Pharmacother 2022; 155:113732. [PMID: 36166964 DOI: 10.1016/j.biopha.2022.113732] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/15/2022] [Accepted: 09/19/2022] [Indexed: 11/24/2022] Open
Abstract
NPC1L1 is a crucial protein involved in sterol lipid absorption and has been shown to play an important role in intestinal cholesterol absorption. Hypercholesterolemia is a significant risk factor for cardiovascular diseases such as coronary heart disease. Screening of NPC1L1 inhibitors is critical for gaining a full understanding of lipid metabolism, developing new cholesterol-lowering medicines, and treating cardiovascular diseases. This work summarized existing methodologies for screening NPC1L1 inhibitors and evaluated their challenges, and will assist the development of novel cholesterol-lowering medications and therapeutic strategies for hypercholesterolemia and other cholesterol-related metabolic disorders.
Collapse
|
9
|
Abstract
PURPOSE OF REVIEW The transintestinal cholesterol efflux (TICE) pathway is the second described route for plasma cholesterol fecal elimination. This article summarizes recent TICE research progresses, involving TICE inducers, molecular determinants of this pathway, and its role in lipoprotein metabolism. RECENT FINDINGS TICE is an active pathway in mice, rats, and humans. Kinetic measurements showed that under basal conditions, the relative contribution of TICE in fecal elimination of plasma cholesterol is quantitatively less important than the hepatobiliary pathway. However, the amplitude of TICE can be induced by numerous nutritional factors and pharmacological drugs. More importantly, by contrast with the stimulation of biliary cholesterol excretion that is associated with an increased risk of gallstone formation, TICE appears as a safer therapeutical target. Finally, several independent studies have demonstrated that TICE is actively contributing to the anti-atherogenic reverse cholesterol pathway reinforcing the interest to better understand its mode of action. The discovery of TICE and the understanding of its mode of action open new therapeutical perspectives for patients at high risk of cardiovascular diseases.
Collapse
|
10
|
The Effects of Anthocyanin-Rich Bilberry Extract on Transintestinal Cholesterol Excretion. Foods 2021; 10:foods10112852. [PMID: 34829135 PMCID: PMC8624570 DOI: 10.3390/foods10112852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/13/2021] [Accepted: 11/15/2021] [Indexed: 11/17/2022] Open
Abstract
Hypercholesterolemia is one of the modifiable and primary risk factors for cardiovascular diseases (CVD). Emerging evidence suggests the stimulation of transintestinal cholesterol excretion (TICE), the nonbiliary cholesterol excretion, using natural products can be an effective way to reduce CVD. Bilberry (Vaccinium myrtillus L.) has been reported to have cardioprotective effects by ameliorating oxidative stress, inflammation, and dyslipidemia. However, the role of bilberry in intestinal cholesterol metabolism is not well understood. To examine the effects of bilberry in intestinal cholesterol metabolism, we measured the genes for cholesterol flux and de novo synthesis in anthocyanin-rich bilberry extract (BE)-treated Caco-2 cells. BE significantly decreased the genes for cholesterol absorption, i.e., Niemann-Pick C1 Like 1 and ATP-binding cassette transporter A1 (ABCA1). In contrast, BE significantly upregulated ABCG8, the apical transporter for cholesterol. There was a significant induction of low-density lipoprotein receptors, with a concomitant increase in cellular uptake of cholesterol in BE-treated cells. The expression of genes for lipogenesis and sirtuins was altered by BE treatment. In the present study, BE altered the genes for cholesterol flux from basolateral to the apical membrane of enterocytes, potentially stimulating TICE. These results support the potential of BE in the prevention of hypercholesterolemia.
Collapse
|
11
|
Tanaka Y, Kamisako T. Regulation of the expression of cholesterol transporters by lipid-lowering drugs ezetimibe and pemafibrate in rat liver and intestine. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166215. [PMID: 34265370 DOI: 10.1016/j.bbadis.2021.166215] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 07/07/2021] [Accepted: 07/08/2021] [Indexed: 12/15/2022]
Abstract
Ezetimibe and pemafibrate are lipid-lowering drugs and promote reverse cholesterol transport. However, it is unknown whether cholesterol is mainly excreted by hepatobiliary excretion or by non-biliary transintestinal cholesterol efflux (TICE). We evaluated the effects of ezetimibe and pemafibrate on hepatic and intestinal cholesterol transporter regulation in Sham-operated rats, and examined the effects of these drugs on TICE in bile duct-ligated rats. Seven-week-old male Sprague-Dawley rats were treated as follows for two weeks: 1) Sham, Sham operation; 2) BDL, bile duct ligation; 3) E-Sham, Sham + ezetimibe; 4) E-BDL, BDL + ezetimibe; 5) P-Sham, Sham + pemafibrate; and 6) P-BDL, BDL + pemafibrate. Blood, liver, jejunum, and feces were collected 72 h post-surgery. Hepatic cholesterol levels were decreased in P-Sham and E-Sham, and were lower in E-BDL and P-BDL than in BDL. Fecal cholesterol levels increased in E-Sham and P-Sham compared with Sham, and were higher in E-BDL and P-BDL than in BDL. In liver, Abcg5 mRNA showed induction in E-Sham, Abcg5 and Abca1 mRNA were induced in P-Sham, Abcg5 mRNA was reduced in E-BDL, and Abca1 mRNA was increased in P-BDL. In jejunum, Abcg5 mRNA was induced in E-Sham. Abcg8 mRNA was induced in E-Sham and P-Sham. NPC1L1 mRNA showed reduced expression in P-Sham and P-BDL. SR-B1 mRNA was reduced in P-Sham, and the expression decreased in P-BDL. LDL receptor mRNA was induced in BDL and P-BDL. Ezetimibe and pemafibrate may promote TICE by increasing Abcg5/g8, while pemafibrate may inhibit intestinal cholesterol absorption by decreasing SR-B1 and NPC1L1.
Collapse
Affiliation(s)
- Yuji Tanaka
- Department of Clinical Laboratory Medicine, Kindai University Faculty of Medicine, Osakasayama, Osaka 589-8511, Japan.
| | - Toshinori Kamisako
- Department of Clinical Laboratory Medicine, Kindai University Faculty of Medicine, Osakasayama, Osaka 589-8511, Japan
| |
Collapse
|
12
|
Inokuchi JI, Kanoh H, Inamori KI, Nagafuku M, Nitta T, Fukase K. Homeostatic and pathogenic roles of the GM3 ganglioside. FEBS J 2021; 289:5152-5165. [PMID: 34125497 DOI: 10.1111/febs.16076] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 05/10/2021] [Accepted: 06/14/2021] [Indexed: 12/14/2022]
Abstract
Two decades ago, we achieved molecular cloning of ganglioside GM3 synthase (GM3S; ST3GAL5), the enzyme responsible for initiating biosynthesis of complex gangliosides. The efforts of our research group since then have been focused on clarifying the physiological and pathological roles of gangliosides, particularly GM3. This review summarizes our long-term studies on the roles of GM3 in insulin resistance and adipogenesis in adipose tissues, cholesterol uptake in intestine, and leptin resistance in hypothalamus. We hypothesized that GM3 plays a role in innate immune function of macrophages and demonstrated that molecular species of GM3 with differing acyl-chain structures and modifications functioned as pro- and anti-inflammatory endogenous Toll-like receptor 4 (TLR4) modulators in macrophages. Very-long-chain and α-hydroxy GM3 species enhanced TLR4 activation, whereas long-chain and unsaturated GM3 species counteracted this effect. Lipidomic analyses of serum and adipose tissues revealed that imbalances between such pro- and anti-inflammatory GM3 species promoted progression of metabolic disorders. GM3 thus functions as a physiological regulatory factor controlling the balance between homeostatic and pathological states. Ongoing studies based on these findings will clarify the mechanisms underlying ganglioside-dependent control of energy homeostasis and innate immune responses.
Collapse
Affiliation(s)
- Jin-Ichi Inokuchi
- Division of Glycopathology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Japan.,Core for Medicine and Science Collaborative Research and Education (MS-CORE), Project Research Center for Fundamental Sciences, Osaka University, Japan
| | - Hirotaka Kanoh
- Division of Glycopathology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Kei-Ichiro Inamori
- Division of Glycopathology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Masakazu Nagafuku
- Division of Glycopathology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Takahiro Nitta
- Division of Glycopathology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Koichi Fukase
- Core for Medicine and Science Collaborative Research and Education (MS-CORE), Project Research Center for Fundamental Sciences, Osaka University, Japan.,Department of Chemistry, Graduate School of Science, Osaka University, Japan
| |
Collapse
|
13
|
Induction of fecal cholesterol excretion is not effective for the treatment of hyperbilirubinemia in Gunn rats. Pediatr Res 2021; 89:510-517. [PMID: 32357361 DOI: 10.1038/s41390-020-0926-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 02/28/2020] [Accepted: 04/01/2020] [Indexed: 11/08/2022]
Abstract
BACKGROUND Unconjugated hyperbilirubinemia, a feature of neonatal jaundice or Crigler-Najjar syndrome, can lead to neurotoxicity and even death. We previously demonstrated that unconjugated bilirubin (UCB) can be eliminated via transintestinal excretion in Gunn rats, a model of unconjugated hyperbilirubinemia, and that this is stimulated by enhancing fecal fatty acid excretion. Since transintestinal excretion also occurs for cholesterol (TICE), we hypothesized that increasing fecal cholesterol excretion and/or TICE could also enhance fecal UCB disposal and subsequently lower plasma UCB concentrations. METHODS To determine whether increasing fecal cholesterol excretion could ameliorate unconjugated hyperbilirubinemia, we treated hyperbilirubinemic Gunn rats with ezetimibe (EZE), an intestinal cholesterol absorption inhibitor, and/or a liver X receptor (LXR) and farnesoid X receptor (FXR) agonist (T0901317 (T09) and obeticholic acid (OCA), respectively), known to stimulate TICE. RESULTS We found that EZE treatment alone or in combination with T09 or OCA increased fecal cholesterol disposal but did not lower plasma UCB levels. CONCLUSIONS These findings do not support a link between the regulation of transintestinal excretion of cholesterol and bilirubin. Furthermore, induction of fecal cholesterol excretion is not a potential therapy for unconjugated hyperbilirubinemia. IMPACT Increasing fecal cholesterol excretion is not effective to treat unconjugated hyperbilirubinemia. This is the first time a potential relation between transintestinal excretion of cholesterol and unconjugated bilirubin is investigated. Transintestinal excretion of cholesterol and unconjugated bilirubin do not seem to be quantitatively linked. Unlike intestinal fatty acids, cholesterol cannot "capture" unconjugated bilirubin to increase its excretion. These results add to our understanding of ways to improve and factors regulating unconjugated bilirubin disposal in hyperbilirubinemic conditions.
Collapse
|
14
|
Nakano T, Inoue I, Takenaka Y, Ito R, Kotani N, Sato S, Nakano Y, Hirasaki M, Shimada A, Murakoshi T. Ezetimibe impairs transcellular lipid trafficking and induces large lipid droplet formation in intestinal absorptive epithelial cells. Biochim Biophys Acta Mol Cell Biol Lipids 2020; 1865:158808. [PMID: 32860884 DOI: 10.1016/j.bbalip.2020.158808] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 07/30/2020] [Accepted: 08/18/2020] [Indexed: 01/26/2023]
Abstract
Ezetimibe inhibits Niemann-Pick C1-like 1 (NPC1L1) protein, which mediates intracellular cholesterol trafficking from the brush border membrane to the endoplasmic reticulum, where chylomicron assembly takes place in enterocytes or in the intestinal absorptive epithelial cells. Cholesterol is a minor lipid constituent of chylomicrons; however, whether or not a shortage of cholesterol attenuates chylomicron assembly is unknown. The aim of this study was to examine the effect of ezetimibe, a potent NPC1L1 inhibitor, on trans-epithelial lipid transport, and chylomicron assembly and secretion in enterocytes. Caco-2 cells, an absorptive epithelial model, grown onto culture inserts were given lipid micelles from the apical side, and chylomicron-like triacylglycerol-rich lipoprotein secreted basolaterally were analyzed after a 24-h incubation period in the presence of ezetimibe up to 50 μM. The secretion of lipoprotein and apolipoprotein B48 were reduced by adding ezetimibe (30% and 34%, respectively). Although ezetimibe allowed the cells to take up cholesterol normally, the esterification was abolished. Meanwhile, oleic acid esterification was unaffected. Moreover, ezetimibe activated sterol regulatory element-binding protein 2 by approximately 1.5-fold. These results suggest that ezetimibe limited cellular cholesterol mobilization required for lipoprotein assembly. In such conditions, large lipid droplet formation in Caco-2 cells and the enterocytes of mice were induced, implying that unprocessed triacylglycerol was sheltered in these compartments. Although ezetimibe did not reduce the post-prandial lipid surge appreciably in triolein-infused mice, the results of the present study indicated that pharmacological actions of ezetimibe may participate in a novel regulatory mechanism for the efficient chylomicron assembly and secretion.
Collapse
Affiliation(s)
- Takanari Nakano
- Department of Biochemistry, Faculty of Medicine, Saitama Medical University, Saitama, Japan.
| | - Ikuo Inoue
- Department of Diabetes and Endocrinology, Faculty of Medicine, Saitama Medical University, Saitama, Japan
| | - Yasuhiro Takenaka
- Department of Diabetes and Endocrinology, Faculty of Medicine, Saitama Medical University, Saitama, Japan; Department of Physiology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Rina Ito
- Department of Biochemistry, Faculty of Medicine, Saitama Medical University, Saitama, Japan
| | - Norihiro Kotani
- Department of Biochemistry, Faculty of Medicine, Saitama Medical University, Saitama, Japan
| | - Sawako Sato
- Department of Diabetes and Endocrinology, Faculty of Medicine, Saitama Medical University, Saitama, Japan
| | - Yuka Nakano
- Department of Diabetes and Endocrinology, Faculty of Medicine, Saitama Medical University, Saitama, Japan
| | - Masataka Hirasaki
- Division of Developmental Biology, Research Center for Genomic Medicine, Faculty of Medicine, Saitama Medical University, Saitama, Japan
| | - Akira Shimada
- Department of Diabetes and Endocrinology, Faculty of Medicine, Saitama Medical University, Saitama, Japan
| | - Takayuki Murakoshi
- Department of Biochemistry, Faculty of Medicine, Saitama Medical University, Saitama, Japan
| |
Collapse
|
15
|
Feng S, Belwal T, Li L, Limwachiranon J, Liu X, Luo Z. Phytosterols and their derivatives: Potential health‐promoting uses against lipid metabolism and associated diseases, mechanism, and safety issues. Compr Rev Food Sci Food Saf 2020; 19:1243-1267. [DOI: 10.1111/1541-4337.12560] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 03/19/2020] [Accepted: 03/24/2020] [Indexed: 12/12/2022]
Affiliation(s)
- Simin Feng
- College of Food Science and TechnologyZhejiang University of Technology Hangzhou 310014 People's Republic of China
- College of Biosystems Engineering and Food Science, Key Laboratory of Agro‐Products Postharvest Handling Ministry of Agriculture, Zhejiang Key Laboratory for Agri‐Food Processing, National‐Local Joint Engineering Laboratory of Intelligent Food Technology and EquipmentZhejiang University Hangzhou 310058 People's Republic of China
- Key Laboratory of Food Macromolecular Resources Processing Technology Research, China National Light IndustryZhejiang University of Technology Hangzhou 310014 People's Republic of China
| | - Tarun Belwal
- College of Biosystems Engineering and Food Science, Key Laboratory of Agro‐Products Postharvest Handling Ministry of Agriculture, Zhejiang Key Laboratory for Agri‐Food Processing, National‐Local Joint Engineering Laboratory of Intelligent Food Technology and EquipmentZhejiang University Hangzhou 310058 People's Republic of China
| | - Li Li
- College of Biosystems Engineering and Food Science, Key Laboratory of Agro‐Products Postharvest Handling Ministry of Agriculture, Zhejiang Key Laboratory for Agri‐Food Processing, National‐Local Joint Engineering Laboratory of Intelligent Food Technology and EquipmentZhejiang University Hangzhou 310058 People's Republic of China
| | - Jarukitt Limwachiranon
- College of Biosystems Engineering and Food Science, Key Laboratory of Agro‐Products Postharvest Handling Ministry of Agriculture, Zhejiang Key Laboratory for Agri‐Food Processing, National‐Local Joint Engineering Laboratory of Intelligent Food Technology and EquipmentZhejiang University Hangzhou 310058 People's Republic of China
| | - Xingquan Liu
- School of Agriculture and Food SciencesZhejiang Agriculture and Forestry University Hangzhou 311300 People's Republic of China
| | - Zisheng Luo
- College of Biosystems Engineering and Food Science, Key Laboratory of Agro‐Products Postharvest Handling Ministry of Agriculture, Zhejiang Key Laboratory for Agri‐Food Processing, National‐Local Joint Engineering Laboratory of Intelligent Food Technology and EquipmentZhejiang University Hangzhou 310058 People's Republic of China
- Ningbo Research InstituteZhejiang University Ningbo 315100 People's Republic of China
- Fuli Institute of Food ScienceZhejiang University Hangzhou 310058 People's Republic of China
| |
Collapse
|
16
|
Hiebl V, Schachner D, Ladurner A, Heiss EH, Stangl H, Dirsch VM. Caco-2 Cells for Measuring Intestinal Cholesterol Transport - Possibilities and Limitations. Biol Proced Online 2020; 22:7. [PMID: 32308567 PMCID: PMC7149936 DOI: 10.1186/s12575-020-00120-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 04/01/2020] [Indexed: 12/19/2022] Open
Abstract
Background The human Caco-2 cell line is a common in vitro model of the intestinal epithelial barrier. As the intestine is a major interface in cholesterol turnover and represents a non-biliary pathway for cholesterol excretion, Caco-2 cells are also a valuable model for studying cholesterol homeostasis, including cholesterol uptake and efflux. Currently available protocols are, however, either sketchy or not consistent among different laboratories. Our aim was therefore to generate a collection of optimized protocols, considering the different approaches of the different laboratories and to highlight possibilities and limitations of measuring cholesterol transport with this cell line. Results We developed comprehensive and quality-controlled protocols for the cultivation of Caco-2 cells on filter inserts in a single tight monolayer. A cholesterol uptake as well as a cholesterol efflux assay is described in detail, including suitable positive controls. We further show that Caco-2 cells can be efficiently transfected for luciferase reporter gene assays in order to determine nuclear receptor activation, main transcriptional regulators of cholesterol transporters (ABCA1, ABCB1, ABCG5/8, NPC1L1). Detection of protein and mRNA levels of cholesterol transporters in cells grown on filter inserts can pose challenges for which we highlight essential steps and alternative approaches for consideration. A protocol for viability assays with cells differentiated on filter inserts is provided for the first time. Conclusions The Caco-2 cell line is widely used in the scientific community as model for the intestinal epithelium, although with highly divergent protocols. The herein provided information and protocols can be a common basis for researchers intending to use Caco-2 cells in the context of cellular cholesterol homeostasis.
Collapse
Affiliation(s)
- Verena Hiebl
- 1Department of Pharmacognosy, University of Vienna, Althanstrasse 14, 1090 Vienna, Austria
| | - Daniel Schachner
- 1Department of Pharmacognosy, University of Vienna, Althanstrasse 14, 1090 Vienna, Austria
| | - Angela Ladurner
- 1Department of Pharmacognosy, University of Vienna, Althanstrasse 14, 1090 Vienna, Austria
| | - Elke H Heiss
- 1Department of Pharmacognosy, University of Vienna, Althanstrasse 14, 1090 Vienna, Austria
| | - Herbert Stangl
- 2Institute of Medical Chemistry, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Vienna, Austria
| | - Verena M Dirsch
- 1Department of Pharmacognosy, University of Vienna, Althanstrasse 14, 1090 Vienna, Austria
| |
Collapse
|
17
|
Kiourtzidis M, Kühn J, Schutkowski A, Baur AC, Hirche F, Stangl GI. Inhibition of Niemann-Pick C1-like protein 1 by ezetimibe reduces uptake of deuterium-labeled vitamin D in mice. J Steroid Biochem Mol Biol 2020; 197:105504. [PMID: 31682937 DOI: 10.1016/j.jsbmb.2019.105504] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 09/22/2019] [Accepted: 10/22/2019] [Indexed: 12/17/2022]
Abstract
For a long time, orally ingested vitamin D was assumed to enter the body exclusively via simple passive diffusion. Recent data from in vitro experiments have described Niemann-Pick C1-like protein 1 (Npc1l1) as an important sterol transporter for vitamin D absorption. However, short-term applications of ezetimibe, which inhibits Npc1l1, were not associated with reduced vitamin D uptake in animals and humans. The current study aimed to elucidate the effect of long-term inhibition of Npc1l1 by ezetimibe on the uptake and storage of orally administered triple deuterated vitamin D3 (vitamin D3-d3). Therefore, 30 male wild-type mice were randomly assigned into three groups and received diets with 25 μg/kg of vitamin D3-d3 that contained 0 (control group), 50 or 100 mg/kg ezetimibe for six weeks. Mice fed diets with 50 or 100 mg/kg ezetimibe had lower circulating levels of cholesterol than control mice (-12 %, -15 %, P < 0.01). In contrast, the concentrations of 7-dehydrocholesterol in serum (P < 0.001) and liver (P < 0.05) were higher in mice treated with ezetimibe than in control mice, indicating an increased sterol synthesis to compensate for cholesterol reduction. Long-term application of ezetimibe significantly reduced the concentrations of vitamin D3-d3 in the serum and tissues of mice. The magnitude of vitamin D3 reduction was comparable between the two ezetimibe groups. In comparison to the control group, mice treated with ezetimibe had lower concentrations of deuterated vitamin D3 compared with the control group in serum (62 %, P < 0.001), liver (79 %, P < 0.001), kidney (54 %, P < 0.001), adipose tissues (55 %, P < 0.001) and muscle (41 %, P < 0.001). Surprisingly, the serum concentration of deuterated 25-hydroxyvitamin D3 was higher in the group fed 100 mg/kg ezetimibe than in the control group (P < 0.05). The protein expression of the vitamin D hydroxylases Cyp2r1, Cyp27a1, Cyp3a11, Cyp24a1 and Cyp2j3 in liver and Cyp27b1 and Cyp24a1 in kidney remained largely unaffected by ezetimibe. To conclude, Npc1l1 appears to be crucial for the uptake of orally ingested vitamin D because long-term inhibition of Npc1l1 by ezetimibe strongly reduced the levels of deuterium-labeled vitamin D in the body; the observed rise in deuterated 25-hydroxyvitamin D3 in serum of these mice can not be explained by the expression levels of the key enzymes involved in vitamin D hydroxylation.
Collapse
Affiliation(s)
- Mikis Kiourtzidis
- Institute of Agricultural and Nutritional Sciences, Martin Luther University Halle-Wittenberg, Germany.
| | - Julia Kühn
- Institute of Agricultural and Nutritional Sciences, Martin Luther University Halle-Wittenberg, Germany.
| | - Alexandra Schutkowski
- Institute of Agricultural and Nutritional Sciences, Martin Luther University Halle-Wittenberg, Germany.
| | - Anja C Baur
- Institute of Agricultural and Nutritional Sciences, Martin Luther University Halle-Wittenberg, Germany; Competence Cluster of Cardiovascular Health and Nutrition (nutriCARD), Halle-Jena-Leipzig, Germany.
| | - Frank Hirche
- Institute of Agricultural and Nutritional Sciences, Martin Luther University Halle-Wittenberg, Germany.
| | - Gabriele I Stangl
- Institute of Agricultural and Nutritional Sciences, Martin Luther University Halle-Wittenberg, Germany; Competence Cluster of Cardiovascular Health and Nutrition (nutriCARD), Halle-Jena-Leipzig, Germany.
| |
Collapse
|
18
|
Strilchuk L, Tocci G, Fogacci F, Cicero AFG. An overview of rosuvastatin/ezetimibe association for the treatment of hypercholesterolemia and mixed dyslipidemia. Expert Opin Pharmacother 2020; 21:531-539. [DOI: 10.1080/14656566.2020.1714028] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Larysa Strilchuk
- Department of Therapy and Medical Diagnostics, Lviv National Medical University, Lviv, Ukraine
| | - Giuliano Tocci
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, University of Rome Sapienza, Sant’Andrea Hospital, Rome, Italy
- Cardiology Unit, IRCCS Neuromed, Pozzilli, Italy
| | - Federica Fogacci
- Medical and Surgical Sciences Department, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Arrigo F. G. Cicero
- Medical and Surgical Sciences Department, Alma Mater Studiorum University of Bologna, Bologna, Italy
| |
Collapse
|
19
|
Lifsey HC, Kaur R, Thompson BH, Bennett L, Temel RE, Graf GA. Stigmasterol stimulates transintestinal cholesterol excretion independent of liver X receptor activation in the small intestine. J Nutr Biochem 2019; 76:108263. [PMID: 31759199 DOI: 10.1016/j.jnutbio.2019.108263] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 10/14/2019] [Accepted: 10/29/2019] [Indexed: 02/09/2023]
Abstract
Despite advances in healthcare, cardiovascular disease (CVD) remains the leading cause of death in the United States. Elevated levels of plasma cholesterol are highly predictive of CVD and stroke and are the principal driver of atherosclerosis. Unfortunately, current cholesterol lowering agents, such as statins, are not known to reverse atherosclerotic disease once it has been established. In preclinical models, agonists of nuclear receptor, LXR, have been shown to reduce and reverse atherosclerosis. Phytosterols are bioactive non-cholesterol sterols that act as LXR agonists and regulate cholesterol metabolism and transport. We hypothesized that stigmasterol would act as an LXR agonist and alter intestinal cholesterol secretion to promote cholesterol elimination. Mice were fed a control diet, or a diet supplemented with stigmasterol (0.3% w/w) or T0901317 (0.015% w/w), a known LXR agonist. In this experiment we analyzed the sterol content of bile, intestinal perfusate, plasma, and feces. Additionally, the liver and small intestine were analyzed for relative levels of transcripts known to be regulated by LXR. We observed that T0901317 robustly promoted cholesterol elimination and acted as a strong LXR agonist. Stigmasterol promoted transintestinal cholesterol secretion through an LXR-independent pathway.
Collapse
Affiliation(s)
| | - Rupinder Kaur
- Department of Pharmaceutical Sciences, College of Pharmacy
| | | | - Lisa Bennett
- Department of Pharmaceutical Sciences, College of Pharmacy
| | - Ryan E Temel
- Department of Physiology, College of Medicine, University of Kentucky; Saha Cardiovascular Research Center
| | - Gregory A Graf
- Department of Pharmaceutical Sciences, College of Pharmacy; Saha Cardiovascular Research Center; Barnstable Brown Diabetes and Obesity Center.
| |
Collapse
|
20
|
van de Peppel IP, Bertolini A, van Dijk TH, Groen AK, Jonker JW, Verkade HJ. Efficient reabsorption of transintestinally excreted cholesterol is a strong determinant for cholesterol disposal in mice. J Lipid Res 2019; 60:1562-1572. [PMID: 31324653 PMCID: PMC6718438 DOI: 10.1194/jlr.m094607] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 07/19/2019] [Indexed: 11/20/2022] Open
Abstract
Transintestinal cholesterol excretion (TICE) is a major route for eliminating cholesterol from the body and a potential therapeutic target for hypercholesterolemia. The underlying mechanism, however, is largely unclear, and its contribution to cholesterol disposal from the body is obscured by the counteracting process of intestinal cholesterol reabsorption. To determine the quantity of TICE independent from its reabsorption, we studied two models of decreased intestinal cholesterol absorption. Cholesterol absorption was inhibited either by ezetimibe or, indirectly, by the genetic inactivation of the intestinal apical sodium-dependent bile acid transporter (ASBT; SLC10A2). Both ezetimibe treatment and Asbt inactivation virtually abrogated fractional cholesterol absorption (from 46% to 4% and 6%, respectively). In both models, fecal neutral sterol excretion and net intestinal cholesterol balance were considerably higher than in control mice (5- and 7-fold, respectively), suggesting that, under physiological conditions, TICE is largely reabsorbed. In addition, the net intestinal cholesterol balance was increased to a similar extent but was not further increased when the models were combined, suggesting that the effect on cholesterol reabsorption was already maximal under either condition alone. On the basis of these findings, we hypothesize that the inhibition of cholesterol (re)absorption combined with stimulating TICE will be most effective in increasing cholesterol disposal.
Collapse
Affiliation(s)
- Ivo P van de Peppel
- Section of Molecular Metabolism and Nutrition, Department of Pediatrics,University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Anna Bertolini
- Section of Molecular Metabolism and Nutrition, Department of Pediatrics,University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Theo H van Dijk
- Department of Laboratory Medicine University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Albert K Groen
- Section of Molecular Metabolism and Nutrition, Department of Pediatrics,University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Laboratory of Experimental Vascular Medicine University of Amsterdam, Academic Medical Center, Amsterdam, The Netherlands
| | - Johan W Jonker
- Section of Molecular Metabolism and Nutrition, Department of Pediatrics,University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| | - Henkjan J Verkade
- Section of Molecular Metabolism and Nutrition, Department of Pediatrics,University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| |
Collapse
|
21
|
Grefhorst A, Verkade HJ, Groen AK. The TICE Pathway: Mechanisms and Lipid-Lowering Therapies. Methodist Debakey Cardiovasc J 2019; 15:70-76. [PMID: 31049152 DOI: 10.14797/mdcj-15-1-70] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Besides the well-known hepatobiliary pathway of cholesterol excretion into the feces, transintestinal cholesterol excretion (TICE) is a second major pathway through which cholesterol is disposed from the body. In the process of TICE, cholesterol is taken up from lipoprotein particles at the basolateral side of the enterocyte and translocates towards the apical side of the enterocyte. At the apical side, the ATP-binding cassette transporters G5 and G8 form a heterodimer that transports cholesterol into the intestinal lumen. A substantial amount of the secreted cholesterol is likely reabsorbed by the cholesterol influx transporter Niemann-Pick C1-Like 1 (NPC1L1) since recent data indicate that inhibition of NPC1L1 increases the efficacy of TICE for disposal of cholesterol via the feces. The pathways and proteins involved in intracellular cholesterol trafficking in the enterocyte have not yet been identified. Therefore, in addition to discussing known mediators of TICE, this review will also examine potential candidates involved in cholesterol translocation in the enterocyte. Both the cholesterol reuptake and efflux pathways can be influenced by pharmaceutical means; thus, the TICE pathway is a very attractive target to increase cholesterol excretion from the body and prevent or mitigate atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Aldo Grefhorst
- AMSTERDAM UNIVERSITY MEDICAL CENTERS, AMSTERDAM, THE NETHERLANDS
| | - Henkjan J Verkade
- UNIVERSITY MEDICAL CENTER GRONINGEN, UNIVERSITY OF GRONINGEN, GRONINGEN, THE NETHERLANDS
| | - Albert K Groen
- AMSTERDAM UNIVERSITY MEDICAL CENTERS, AMSTERDAM, THE NETHERLANDS.,UNIVERSITY MEDICAL CENTER GRONINGEN, UNIVERSITY OF GRONINGEN, GRONINGEN, THE NETHERLANDS
| |
Collapse
|
22
|
Plat J, Baumgartner S, Vanmierlo T, Lütjohann D, Calkins KL, Burrin DG, Guthrie G, Thijs C, Te Velde AA, Vreugdenhil ACE, Sverdlov R, Garssen J, Wouters K, Trautwein EA, Wolfs TG, van Gorp C, Mulder MT, Riksen NP, Groen AK, Mensink RP. Plant-based sterols and stanols in health & disease: "Consequences of human development in a plant-based environment?". Prog Lipid Res 2019; 74:87-102. [PMID: 30822462 DOI: 10.1016/j.plipres.2019.02.003] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Revised: 02/13/2019] [Accepted: 02/25/2019] [Indexed: 01/27/2023]
Abstract
Dietary plant sterols and stanols as present in our diet and in functional foods are well-known for their inhibitory effects on intestinal cholesterol absorption, which translates into lower low-density lipoprotein cholesterol concentrations. However, emerging evidence suggests that plant sterols and stanols have numerous additional health effects, which are largely unnoticed in the current scientific literature. Therefore, in this review we pose the intriguing question "What would have occurred if plant sterols and stanols had been discovered and embraced by disciplines such as immunology, hepatology, pulmonology or gastroenterology before being positioned as cholesterol-lowering molecules?" What would then have been the main benefits and fields of application of plant sterols and stanols today? We here discuss potential effects ranging from its presence and function intrauterine and in breast milk towards a potential role in the development of non-alcoholic steatohepatitis (NASH), cardiovascular disease (CVD), inflammatory bowel diseases (IBD) and allergic asthma. Interestingly, effects clearly depend on the route of entrance as observed in intestinal-failure associated liver disease (IFALD) during parenteral nutrition regimens. It is only until recently that effects beyond lowering of cholesterol concentrations are being explored systematically. Thus, there is a clear need to understand the full health effects of plant sterols and stanols.
Collapse
Affiliation(s)
- J Plat
- Department of Nutrition and Movement Sciences, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, the Netherlands.
| | - S Baumgartner
- Department of Nutrition and Movement Sciences, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, the Netherlands
| | - T Vanmierlo
- Department of Immunology and Biochemistry, Biomedical Research Institute (Biomed) Hasselt University, Hasselt, Belgium; Division of Translational Neuroscience, Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, the Netherlands
| | - D Lütjohann
- Institute of Clinical Chemistry and Clinical Pharmacology, University of Bonn, Bonn, Germany
| | - K L Calkins
- David Geffen School of Medicine, University of California Los Angeles, Mattel Children's Hospital at UCLA, Los Angeles, CA; Department of Pediatrics, Division of Neonatology and Developmental Biology, Neonatal Research Center, USA
| | - D G Burrin
- Department of Pediatrics, USDA Children's Nutrition Research Center, Baylor College of Medicine, Houston, USA
| | - G Guthrie
- Department of Pediatrics, USDA Children's Nutrition Research Center, Baylor College of Medicine, Houston, USA
| | - C Thijs
- Department of Epidemiology, Care and Public Health Research Institute (CAPHRI), Maastricht University, Maastricht, the Netherlands
| | - A A Te Velde
- Tytgat Institute for Liver and Intestinal Research, Amsterdam Medical Center, the Netherlands
| | - A C E Vreugdenhil
- Department of Pediatrics, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, the Netherlands
| | - R Sverdlov
- Department of Molecular Genetics, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, the Netherlands
| | - J Garssen
- Utrecht University, Division Pharmacology, Utrecht Institute for Pharmaceutical Sciences, the Netherlands
| | - K Wouters
- Department of Internal Medicine, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | | | - T G Wolfs
- Department of Pediatrics, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, the Netherlands
| | - C van Gorp
- Department of Pediatrics, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, the Netherlands
| | - M T Mulder
- Department of Internal Medicine, Rotterdam University, Rotterdam, the Netherlands
| | - N P Riksen
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - A K Groen
- Amsterdam Diabetes Center and Department of Vascular Medicine, Academic Medical Center, Amsterdam, the Netherlands
| | - R P Mensink
- Department of Nutrition and Movement Sciences, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
23
|
Nakano T, Inoue I, Murakoshi T. A Newly Integrated Model for Intestinal Cholesterol Absorption and Efflux Reappraises How Plant Sterol Intake Reduces Circulating Cholesterol Levels. Nutrients 2019; 11:nu11020310. [PMID: 30717222 PMCID: PMC6412963 DOI: 10.3390/nu11020310] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 01/28/2019] [Accepted: 01/29/2019] [Indexed: 12/27/2022] Open
Abstract
Cholesterol homeostasis is maintained through a balance of de novo synthesis, intestinal absorption, and excretion from the gut. The small intestine contributes to cholesterol homeostasis by absorbing and excreting it, the latter of which is referred to as trans-intestinal cholesterol efflux (TICE). Because the excretion efficiency of endogenous cholesterol is inversely associated with the development of atherosclerosis, TICE provides an attractive therapeutic target. Thus, elucidation of the mechanism is warranted. We have shown that intestinal cholesterol absorption and TICE are inversely correlated in intestinal perfusion experiments in mice. In this review, we summarized 28 paired data sets for absorption efficiency and fecal neutral sterol excretion, a surrogate marker of TICE, obtained from 13 available publications in a figure, demonstrating the inverse correlation were nearly consistent with the assumption. We then offer a bidirectional flux model that accommodates absorption and TICE occurring in the same segment. In this model, the brush border membrane (BBM) of intestinal epithelial cells stands as the dividing ridge for cholesterol fluxes, making the opposite fluxes competitive and being coordinated by shared BBM-localized transporters, ATP-binding cassette G5/G8 and Niemann-Pick C1-like 1. Furthermore, the idea is applied to address how excess plant sterol/stanol (PS) intake reduces circulating cholesterol level, because the mechanism is still unclear. We propose that unabsorbable PS repeatedly shuttles between the BBM and lumen and promotes concomitant cholesterol efflux. Additionally, PSs, which are chemically analogous to cholesterol, may disturb the trafficking machineries that transport cholesterol to the cell interior.
Collapse
Affiliation(s)
- Takanari Nakano
- Department of Biochemistry, Faculty of Medicine, Saitama Medical University, Saitama 350-0495, Japan.
| | - Ikuo Inoue
- Department of Diabetes and Endocrinology, Faculty of Medicine, Saitama Medical University, Saitama 350-0495, Japan.
| | - Takayuki Murakoshi
- Department of Biochemistry, Faculty of Medicine, Saitama Medical University, Saitama 350-0495, Japan.
| |
Collapse
|
24
|
van Zutphen T, Bertolini A, de Vries HD, Bloks VW, de Boer JF, Jonker JW, Kuipers F. Potential of Intestine-Selective FXR Modulation for Treatment of Metabolic Disease. Handb Exp Pharmacol 2019; 256:207-234. [PMID: 31236687 DOI: 10.1007/164_2019_233] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Farnesoid X receptor controls bile acid metabolism, both in the liver and intestine. This potent nuclear receptor not only maintains homeostasis of its own ligands, i.e., bile acids, but also regulates glucose and lipid metabolism as well as the immune system. These findings have led to substantial interest for FXR as a therapeutic target and to the recent approval of an FXR agonist for treating primary biliary cholangitis as well as ongoing clinical trials for other liver diseases. Given that FXR biology is complex, including moderate expression in tissues outside of the enterohepatic circulation, temporal expression of isoforms, posttranscriptional modifications, and the existence of several other bile acid-responsive receptors such as TGR5, clinical application of FXR modulators warrants thorough understanding of its actions. Recent findings have demonstrated remarkable physiological effects of targeting FXR specifically in the intestine (iFXR), thereby avoiding systemic release of modulators. These include local effects such as improvement of intestinal barrier function and intestinal cholesterol turnover, as well as systemic effects such as improvements in glucose homeostasis, insulin sensitivity, and nonalcoholic fatty liver disease (NAFLD). Intriguingly, metabolic improvements have been observed with both an iFXR agonist that leads to production of enteric Fgf15 and increased energy expenditure in adipose tissues and antagonists by reducing systemic ceramide levels and hepatic glucose production. Here we review the recent findings on the role of intestinal FXR and its targeting in metabolic disease.
Collapse
Affiliation(s)
- Tim van Zutphen
- Department of Pediatrics, University Medical Center Groningen, Faculty Campus Fryslân, University of Groningen, Groningen, The Netherlands
- University of Groningen, Leeuwarden, The Netherlands
| | - Anna Bertolini
- Department of Pediatrics, University Medical Center Groningen, Faculty Campus Fryslân, University of Groningen, Groningen, The Netherlands
| | - Hilde D de Vries
- Department of Pediatrics, University Medical Center Groningen, Faculty Campus Fryslân, University of Groningen, Groningen, The Netherlands
- University of Groningen, Leeuwarden, The Netherlands
| | - Vincent W Bloks
- Department of Pediatrics, University Medical Center Groningen, Faculty Campus Fryslân, University of Groningen, Groningen, The Netherlands
| | - Jan Freark de Boer
- Department of Pediatrics, University Medical Center Groningen, Faculty Campus Fryslân, University of Groningen, Groningen, The Netherlands
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Johan W Jonker
- Department of Pediatrics, University Medical Center Groningen, Faculty Campus Fryslân, University of Groningen, Groningen, The Netherlands
| | - Folkert Kuipers
- Department of Pediatrics, University Medical Center Groningen, Faculty Campus Fryslân, University of Groningen, Groningen, The Netherlands.
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| |
Collapse
|
25
|
Nakano T, Inoue I, Takenaka Y, Ikegami Y, Kotani N, Shimada A, Noda M, Murakoshi T. Luminal plant sterol promotes brush border membrane-to-lumen cholesterol efflux in the small intestine. J Clin Biochem Nutr 2018; 63:102-105. [PMID: 30279620 PMCID: PMC6160726 DOI: 10.3164/jcbn.17-116] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2017] [Accepted: 12/19/2017] [Indexed: 11/22/2022] Open
Abstract
Plant sterols are used as food additives to reduce intestinal cholesterol absorption. They also increase fecal neutral sterol (FNS) excretion irrespective of the absorption inhibition. Intestine-mediated reverse cholesterol transport, or trans-intestinal cholesterol efflux (TICE), provides the major part of the increase of FNS excretion. However, it is unknown whether plant sterols stimulate TICE or not. We have shown previously that TICE can be evaluated by brush border membrane (BBM)-to-lumen cholesterol efflux. Thus, we examined whether luminal plant sterols stimulate BBM-to-lumen cholesterol efflux in the intestinal tract or not in mice. Cannulated upper jejunum that had been pre-labeled with orally given 3H-cholesterol, was flushed and perfused to collect 3H-cholesterol effluxed back into the lumen from the BBM to estimate the efflux efficiency. Adding 0.5 mg/ml of plant sterols, but not cholesterol, in the perfusion solution doubled the efflux. Plant sterols enter the BBM and are effluxed back to the lumen rapidly, in which process cholesterol transporters in the BBM are involved. We thus speculate that phytosterols alter cholesterol flux in the BBM; thereby, increases BBM-to-lumen cholesterol efflux, resulting in the increased TICE.
Collapse
Affiliation(s)
- Takanari Nakano
- Department of Biochemistry, Faculty of Medicine, Saitama Medical University, Iruma, Saitama 350-0495, Japan
| | - Ikuo Inoue
- Department of Diabetes and Endocrinology, Faculty of Medicine, Saitama Medical University, Iruma, Saitama 350-0495, Japan
| | - Yasuhiro Takenaka
- Department of Physiology, Nippon Medical School, Bunkyo-ku, Tokyo 113-8602, Japan
| | - Yuichi Ikegami
- Department of Physiology, Nippon Medical School, Bunkyo-ku, Tokyo 113-8602, Japan
| | - Norihiro Kotani
- Department of Biochemistry, Faculty of Medicine, Saitama Medical University, Iruma, Saitama 350-0495, Japan
| | - Akira Shimada
- Department of Diabetes and Endocrinology, Faculty of Medicine, Saitama Medical University, Iruma, Saitama 350-0495, Japan
| | - Mitsuhiko Noda
- Department of Diabetes and Endocrinology, Faculty of Medicine, Saitama Medical University, Iruma, Saitama 350-0495, Japan
| | - Takayuki Murakoshi
- Department of Biochemistry, Faculty of Medicine, Saitama Medical University, Iruma, Saitama 350-0495, Japan
| |
Collapse
|
26
|
de Boer JF, Kuipers F, Groen AK. Cholesterol Transport Revisited: A New Turbo Mechanism to Drive Cholesterol Excretion. Trends Endocrinol Metab 2018; 29:123-133. [PMID: 29276134 DOI: 10.1016/j.tem.2017.11.006] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2017] [Revised: 11/26/2017] [Accepted: 11/29/2017] [Indexed: 12/31/2022]
Abstract
A fine-tuned balance between cholesterol uptake and excretion by the body is pivotal to maintain health and to remain free from the deleterious consequences of cholesterol accumulation such as cardiovascular disease. The pathways involved in intracellular and extracellular cholesterol transport are a subject of intense investigation and are being unraveled in increasing detail. In addition, insight into the complex interactions between cholesterol and bile acid metabolism has increased considerably in the last couple of years. This review provides an overview of the mechanisms involved in cholesterol uptake and excretion, with a particular emphasis on the most recent progress in this field. Special attention is given to the transintestinal cholesterol excretion (TICE) pathway, which was recently demonstrated to have a remarkably high transport capacity and to be sensitive to pharmacological modulation.
Collapse
Affiliation(s)
- Jan Freark de Boer
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| | - Folkert Kuipers
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Albert K Groen
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Department of Vascular Medicine, University of Amsterdam Academic Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
27
|
Abstract
PURPOSE OF REVIEW To discuss recent insights into the measurement and cellular basis of transintestinal cholesterol excretion (TICE) in humans and to explore TICE as a therapeutic target for increasing reverse cholesterol transport. RECENT FINDINGS TICE is the net effect of cholesterol excretion by the enterocyte into the intestinal lumen and is the balance between input and output fluxes through the enterocytes. These fluxes are: cholesterol excretion into the intestinal lumen mainly via ATP-binding cassette (ABC) G5/8, cholesterol absorption from the intestine by Niemann-Pick C1 like protein 1, the uptake of plasma lipoproteins by enterocytes at the basolateral membrane, and the excretion of cholesterol in chylomicrons into the lymph. Multiple studies have shown that TICE contributes to fecal neutral sterol (FNS) excretion in humans. TICE can be targeted with plant sterols, liver X receptor agonists, bile acids, ezetimibe, and proprotein convertase subtilisin/kexin type 9 inhibitors. SUMMARY TICE contributes significantly to FNS excretion in humans, independently of the biliary pathway. Knowledge about its underlying cellular mechanisms surges through in-vivo and in-vitro studies in mice and humans. TICE might be an interesting therapeutic target for increasing cholesterol disposal with the feces. Albeit multiple therapeutic options are available, studies showing clinical benefit are still needed.
Collapse
Affiliation(s)
| | | | - Albert K Groen
- Department of Experimental Vascular Medicine, Academic Medical Center, Amsterdam
- Department of Pediatrics, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
28
|
Duntas LH, Brenta G. A Renewed Focus on the Association Between Thyroid Hormones and Lipid Metabolism. Front Endocrinol (Lausanne) 2018; 9:511. [PMID: 30233497 PMCID: PMC6129606 DOI: 10.3389/fendo.2018.00511] [Citation(s) in RCA: 117] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 08/15/2018] [Indexed: 12/19/2022] Open
Abstract
Thyroid dysfunction, manifesting as either overt or subclinical hypothyroidism, negatively affects lipid metabolism: this leads to hypercholesterolemia which progressively increases the risk for cardiovascular disease and, potentially, mortality. Hypercholesterolemia in hypothyroidism is mainly due to a reduction in low-density lipoprotein (LDL) receptor activity, this accompanied by concomitant diminishing control by triiodothyronine (T3) of sterol regulatory element-binding protein 2 (SREBP-2), which modulates cholesterol biosynthesis by regulating rate-limit degrading enzyme 3-hydroxy-3-methylglutaryl-coenzyme A reductase (HMG-CoA) activity. Recently, 3,5-diiodothyronine (T2), a natural thyroid hormone derivative, was found to repress the transcription factor carbohydrate-response element-binding protein (ChREBP) and also to be involved in lipid catabolism and lipogenesis, though via a different pathway than that of T3. While thyroid hormone could therapeutically reverse the dyslipidemic profile commonly occurring in hypothyroidism, it should be borne in mind that the potency of the effects may be age-and sex-dependent. Thyroid hormone administration possibly also sustains and enhances the efficacy of hypolipidemic drugs, such as statins, ezetimibe and proprotein convertase subtilisin/kexin type 9 (PCSK9), in patients with dyslipidemia and hypothyroidism.
Collapse
Affiliation(s)
- Leonidas H. Duntas
- Unit of Endocrinology Diabetes and Metabolism, Thyroid Section, Evgenideion Hospital, University of Athens, Athens, Greece
- *Correspondence: Leonidas H. Duntas
| | - Gabriela Brenta
- Unit of Endocrinology and Metabolism, Thyroid Section, Dr. Cesar Milstein Hospital, Buenos Aires, Argentina
| |
Collapse
|
29
|
Mukherjee P, Hough G, Chattopadhyay A, Navab M, Fogelman HR, Meriwether D, Williams K, Bensinger S, Moller T, Faull KF, Lusis AJ, Iruela-Arispe ML, Bostrom KI, Tontonoz P, Reddy ST, Fogelman AM. Transgenic tomatoes expressing the 6F peptide and ezetimibe prevent diet-induced increases of IFN-β and cholesterol 25-hydroxylase in jejunum. J Lipid Res 2017; 58:1636-1647. [PMID: 28592401 PMCID: PMC5538285 DOI: 10.1194/jlr.m076554] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 06/02/2017] [Indexed: 12/20/2022] Open
Abstract
Feeding LDL receptor (LDLR)-null mice a Western diet (WD) increased the expression of IFN-β in jejunum as determined by quantitative RT-PCR (RT-qPCR), immunohistochemistry (IHC), and ELISA (all P < 0.0001). WD also increased the expression of cholesterol 25-hydroxylase (CH25H) as measured by RT-qPCR (P < 0.0001), IHC (P = 0.0019), and ELISA (P < 0.0001), resulting in increased levels of 25-hydroxycholesterol (25-OHC) in jejunum as determined by LC-MS/MS (P < 0.0001). Adding ezetimibe at 10 mg/kg/day or adding a concentrate of transgenic tomatoes expressing the 6F peptide (Tg6F) at 0.06% by weight of diet substantially ameliorated these changes. Adding either ezetimibe or Tg6F to WD also ameliorated WD-induced changes in plasma lipids, serum amyloid A, and HDL cholesterol. Adding the same doses of ezetimibe and Tg6F together to WD (combined formulation) was generally more efficacious compared with adding either agent alone. Surprisingly, adding ezetimibe during the preparation of Tg6F, but before addition to WD, was more effective than the combined formulation for all parameters measured in jejunum (P = 0.0329 to P < 0.0001). We conclude the following: i) WD induces IFN-β, CH25H, and 25-OHC in jejunum; and ii) Tg6F and ezetimibe partially ameliorate WD-induced inflammation by preventing WD-induced increases in IFN-β, CH25H, and 25-OHC.
Collapse
Affiliation(s)
- Pallavi Mukherjee
- Departments of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Greg Hough
- Departments of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Arnab Chattopadhyay
- Departments of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Mohamad Navab
- Departments of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Hannah R Fogelman
- Departments of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - David Meriwether
- Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Kevin Williams
- Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Steven Bensinger
- Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Travis Moller
- Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Kym F Faull
- Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Aldons J Lusis
- Departments of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA; Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA; Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - M Luisa Iruela-Arispe
- Department of Molecular, Cell, and Developmental Biology, College of Letters and Sciences, University of California, Los Angeles, CA
| | - Kristina I Bostrom
- Departments of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Peter Tontonoz
- Howard Hughes Medical Institute, Los Angeles, CA; Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Srinivasa T Reddy
- Departments of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA; Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA; Obstetrics and Gynecology, David Geffen School of Medicine at UCLA, Los Angeles, CA.
| | - Alan M Fogelman
- Departments of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| |
Collapse
|
30
|
Lin X, Racette SB, Ma L, Wallendorf M, Ostlund RE. Ezetimibe Increases Endogenous Cholesterol Excretion in Humans. Arterioscler Thromb Vasc Biol 2017; 37:990-996. [PMID: 28279967 DOI: 10.1161/atvbaha.117.309119] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 02/27/2017] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Ezetimibe improves cardiovascular outcomes when added to optimum statin treatment. It lowers low-density lipoprotein cholesterol and percent intestinal cholesterol absorption, but the exact cardioprotective mechanism is unknown. We tested the hypothesis that the dominant effect of ezetimibe is to increase the reverse transport of cholesterol from rapidly mixing endogenous cholesterol pool into the stool. APPROACH AND RESULTS In a randomized, placebo-controlled, double-blind parallel trial in 24 healthy subjects with low-density lipoprotein cholesterol 100 to 200 mg/dL, we measured cholesterol metabolism before and after a 6-week treatment period with ezetimibe 10 mg/d or placebo. Plasma cholesterol was labeled by intravenous infusion of cholesterol-d7 in a lipid emulsion and dietary cholesterol with cholesterol-d5 and sitostanol-d4 solubilized in oil. Plasma and stool samples collected during a cholesterol- and phytosterol-controlled metabolic kitchen diet were analyzed by mass spectrometry. Ezetimibe reduced intestinal cholesterol absorption efficiency 30±4.3% (SE, P<0.0001) and low-density lipoprotein cholesterol 19.8±1.9% (P=0.0001). Body cholesterol pool size was unchanged, but fecal endogenous cholesterol excretion increased 66.6±12.2% (P<0.0001) and percent cholesterol excretion from body pools into the stool increased 74.7±14.3% (P<0.0001), whereas plasma cholesterol turnover rose 26.2±3.6% (P=0.0096). Fecal bile acids were unchanged. CONCLUSIONS Ezetimibe increased the efficiency of reverse cholesterol transport from rapidly mixing plasma and tissue pools into the stool. Further work is needed to examine the potential relation of reverse cholesterol transport and whole body cholesterol metabolism to coronary events and the treatment of atherosclerosis. CLINICAL TRIALS REGISTRATION URL: http://www.clinicaltrials.gov. Unique identifier: NCT01603758.
Collapse
Affiliation(s)
- Xiaobo Lin
- From the Division of Endocrinology, Metabolism & Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO (X.L., S.B.R., L.M., R.E.O.); Program in Physical Therapy, Washington University School of Medicine, St. Louis, MO (S.B.R.); and Division of Biostatistics, Washington University School of Medicine, St. Louis, MO (M.W.)
| | - Susan B Racette
- From the Division of Endocrinology, Metabolism & Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO (X.L., S.B.R., L.M., R.E.O.); Program in Physical Therapy, Washington University School of Medicine, St. Louis, MO (S.B.R.); and Division of Biostatistics, Washington University School of Medicine, St. Louis, MO (M.W.)
| | - Lina Ma
- From the Division of Endocrinology, Metabolism & Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO (X.L., S.B.R., L.M., R.E.O.); Program in Physical Therapy, Washington University School of Medicine, St. Louis, MO (S.B.R.); and Division of Biostatistics, Washington University School of Medicine, St. Louis, MO (M.W.)
| | - Michael Wallendorf
- From the Division of Endocrinology, Metabolism & Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO (X.L., S.B.R., L.M., R.E.O.); Program in Physical Therapy, Washington University School of Medicine, St. Louis, MO (S.B.R.); and Division of Biostatistics, Washington University School of Medicine, St. Louis, MO (M.W.)
| | - Richard E Ostlund
- From the Division of Endocrinology, Metabolism & Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO (X.L., S.B.R., L.M., R.E.O.); Program in Physical Therapy, Washington University School of Medicine, St. Louis, MO (S.B.R.); and Division of Biostatistics, Washington University School of Medicine, St. Louis, MO (M.W.).
| |
Collapse
|
31
|
The Interpretation of Cholesterol Balance Derived Synthesis Data and Surrogate Noncholesterol Plasma Markers for Cholesterol Synthesis under Lipid Lowering Therapies. CHOLESTEROL 2017; 2017:5046294. [PMID: 28321334 PMCID: PMC5340945 DOI: 10.1155/2017/5046294] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 12/21/2016] [Accepted: 01/26/2017] [Indexed: 12/12/2022]
Abstract
The cholesterol balance procedure allows the calculation of cholesterol synthesis based on the assumption that loss of endogenous cholesterol via fecal excretion and bile acid synthesis is compensated by de novo synthesis. Under ezetimibe therapy hepatic cholesterol is diminished which can be compensated by hepatic de novo synthesis and hepatic extraction of plasma cholesterol. The plasma lathosterol concentration corrected for total cholesterol concentration (R_Lath) as a marker of de novo cholesterol synthesis is increased during ezetimibe treatment but unchanged under treatment with ezetimibe and simvastatin. Cholesterol balance derived synthesis data increase during both therapies. We hypothesize the following. (1) The cholesterol balance data must be applied to the hepatobiliary cholesterol pool. (2) The calculated cholesterol synthesis value is the sum of hepatic de novo synthesis and the net plasma-liver cholesterol exchange rate. (3) The reduced rate of biliary cholesterol absorption is the major trigger for the regulation of hepatic cholesterol metabolism under ezetimibe treatment. Supportive experimental and literature data are presented that describe changes of cholesterol fluxes under ezetimibe, statin, and combined treatments in omnivores and vegans, link plasma R_Lath to liver function, and define hepatic de novo synthesis as target for regulation of synthesis. An ezetimibe dependent direct hepatic drug effect cannot be excluded.
Collapse
|
32
|
Dugardin C, Briand O, Touche V, Schonewille M, Moreau F, Le May C, Groen AK, Staels B, Lestavel S. Retrograde cholesterol transport in the human Caco-2/TC7 cell line: a model to study trans-intestinal cholesterol excretion in atherogenic and diabetic dyslipidemia. Acta Diabetol 2017; 54:191-199. [PMID: 27796655 DOI: 10.1007/s00592-016-0936-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Accepted: 10/18/2016] [Indexed: 12/19/2022]
Abstract
AIMS The dyslipidemia associated with type 2 diabetes is a major risk factor for the development of atherosclerosis. Trans-intestinal cholesterol excretion (TICE) has recently been shown to contribute, together with the classical hepatobiliary route, to fecal cholesterol excretion and cholesterol homeostasis. The aim of this study was to develop an in vitro cell model to investigate enterocyte-related processes of TICE. METHODS Differentiated Caco-2/TC7 cells were grown on transwells and incubated basolaterally (blood side) with human plasma and apically (luminal side) with lipid micelles. Radioactive and fluorescent cholesterol tracers were used to investigate cholesterol uptake at the basolateral membrane, intracellular distribution and apical excretion. RESULTS Our results show that cholesterol is taken up at the basolateral membrane, accumulates intracellularly as lipid droplets and undergoes a cholesterol acceptor-facilitated and progressive excretion through the apical membrane of enterocytes. The overall process is abolished at 4 °C, suggesting a biologically active phenomenon. Moreover, this trans-enterocytic retrograde cholesterol transport displays some TICE features like modulation by PCSK9 and an ABCB1 inhibitor. Finally, we highlight the involvement of microtubules in the transport of plasma cholesterol from basolateral to apical pole of enterocytes. CONCLUSIONS The human Caco-2/TC7 cell line appears a good in vitro model to investigate the enterocytic molecular mechanisms of TICE, which may help to identify intestinal molecular targets to enhance reverse cholesterol transport and fight against dyslipidemia.
Collapse
Affiliation(s)
- Camille Dugardin
- Inserm, CHU Lille, Institut Pasteur de Lille, U1011 - EGID, Univ. Lille, 59000, Lille, France
| | - Olivier Briand
- Inserm, CHU Lille, Institut Pasteur de Lille, U1011 - EGID, Univ. Lille, 59000, Lille, France
| | - Véronique Touche
- Inserm, CHU Lille, Institut Pasteur de Lille, U1011 - EGID, Univ. Lille, 59000, Lille, France
| | - Marleen Schonewille
- University Medical Center Groningen, Department of Pediatrics, University of Groningen, Groningen, The Netherlands
| | | | - Cédric Le May
- INSERM, UMR 1087, CNRS UMR 6291, 44000, Nantes, France
| | - Albert K Groen
- University Medical Center Groningen, Department of Pediatrics, University of Groningen, Groningen, The Netherlands
- Academic Medical Center, Amsterdam, The Netherlands
| | - Bart Staels
- Inserm, CHU Lille, Institut Pasteur de Lille, U1011 - EGID, Univ. Lille, 59000, Lille, France.
| | - Sophie Lestavel
- Inserm, CHU Lille, Institut Pasteur de Lille, U1011 - EGID, Univ. Lille, 59000, Lille, France
| |
Collapse
|
33
|
Jakulj L, van Dijk TH, de Boer JF, Kootte RS, Schonewille M, Paalvast Y, Boer T, Bloks VW, Boverhof R, Nieuwdorp M, Beuers UHW, Stroes ESG, Groen AK. Transintestinal Cholesterol Transport Is Active in Mice and Humans and Controls Ezetimibe-Induced Fecal Neutral Sterol Excretion. Cell Metab 2016; 24:783-794. [PMID: 27818259 DOI: 10.1016/j.cmet.2016.10.001] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Revised: 05/09/2016] [Accepted: 09/30/2016] [Indexed: 01/10/2023]
Abstract
Except for conversion to bile salts, there is no major cholesterol degradation pathway in mammals. Efficient excretion from the body is therefore a crucial element in cholesterol homeostasis. Yet, the existence and importance of cholesterol degradation pathways in humans is a matter of debate. We quantified cholesterol fluxes in 15 male volunteers using a cholesterol balance approach. Ten participants repeated the protocol after 4 weeks of treatment with ezetimibe, an inhibitor of intestinal and biliary cholesterol absorption. Under basal conditions, about 65% of daily fecal neutral sterol excretion was bile derived, with the remainder being contributed by direct transintestinal cholesterol excretion (TICE). Surprisingly, ezetimibe induced a 4-fold increase in cholesterol elimination via TICE. Mouse studies revealed that most of ezetimibe-induced TICE flux is mediated by the cholesterol transporter Abcg5/Abcg8. In conclusion, TICE is active in humans and may serve as a novel target to stimulate cholesterol elimination in patients at risk for cardiovascular disease.
Collapse
Affiliation(s)
- Lily Jakulj
- Department of Vascular Medicine, Academic Medical Center, Amsterdam 1105AZ, the Netherlands
| | - Theo H van Dijk
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen 9713ZG, the Netherlands
| | - Jan Freark de Boer
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen 9713ZG, the Netherlands
| | - Ruud S Kootte
- Department of Vascular Medicine, Academic Medical Center, Amsterdam 1105AZ, the Netherlands
| | - Marleen Schonewille
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen 9713ZG, the Netherlands
| | - Yared Paalvast
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen 9713ZG, the Netherlands
| | - Theo Boer
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen 9713ZG, the Netherlands
| | - Vincent W Bloks
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen 9713ZG, the Netherlands
| | - Renze Boverhof
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen 9713ZG, the Netherlands
| | - Max Nieuwdorp
- Department of Vascular Medicine, Academic Medical Center, Amsterdam 1105AZ, the Netherlands
| | - Ulrich H W Beuers
- Department of Gastroenterology and Hepatology, Academic Medical Center, Amsterdam 1105AZ, the Netherlands
| | - Erik S G Stroes
- Department of Vascular Medicine, Academic Medical Center, Amsterdam 1105AZ, the Netherlands
| | - Albert K Groen
- Department of Vascular Medicine, Academic Medical Center, Amsterdam 1105AZ, the Netherlands; Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen 9713ZG, the Netherlands; Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen 9713ZG, the Netherlands; Amsterdam Diabetes Research Center, Academic Medical Center, Amsterdam 1105AZ, the Netherlands; Groningen Center of Systems Biology, University Medical Center Groningen, Groningen 9713ZG, the Netherlands.
| |
Collapse
|